151
|
Duan JJ, Qiu W, Xu SL, Wang B, Ye XZ, Ping YF, Zhang X, Bian XW, Yu SC. Strategies for isolating and enriching cancer stem cells: well begun is half done. Stem Cells Dev 2013; 22:2221-39. [PMID: 23540661 DOI: 10.1089/scd.2012.0613] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) constitute a subpopulation of cancer cells that have the potential for self-renewal, multipotent differentiation, and tumorigenicity. Studies on CSC biology and CSC-targeted therapies depend on CSC isolation and/or enrichment methodologies. Scientists have conducted extensive research in this field since John Dick's group successfully isolated CSCs based on the expression of the CD34 and CD38 surface markers. Progress in CSC research has been greatly facilitated by the enrichment and isolation of these cells. In this review, we summarize the current strategies used in our and other laboratories for CSC isolation and enrichment, including methods based on stem cell surface markers, intracellular enzyme activity, the concentration of reactive oxygen species, the mitochondrial membrane potential, promoter-driven fluorescent protein expression, autofluorescence, suspension/adherent culture, cell division, the identification of side population cells, resistance to cytotoxic compounds or hypoxia, invasiveness/adhesion, immunoselection, and physical property. Although many challenges remain to be overcome, it is reasonable to believe that more reliable, efficient, and convenient methods will be developed in the near future.
Collapse
Affiliation(s)
- Jiang-Jie Duan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Bu P, Chen KY, Chen JH, Wang L, Walters J, Shin YJ, Goerger JP, Sun J, Witherspoon M, Rakhilin N, Li J, Yang H, Milsom J, Lee S, Zipfel W, Jin MM, Gümüşcedil ZH, Lipkin SM, Shen X. A microRNA miR-34a-regulated bimodal switch targets Notch in colon cancer stem cells. Cell Stem Cell 2013; 12:602-15. [PMID: 23642368 PMCID: PMC3646336 DOI: 10.1016/j.stem.2013.03.002] [Citation(s) in RCA: 290] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 11/25/2012] [Accepted: 03/04/2013] [Indexed: 12/15/2022]
Abstract
microRNAs regulate developmental cell-fate decisions, tissue homeostasis, and oncogenesis in distinct ways relative to proteins. Here, we show that the tumor suppressor microRNA miR-34a is a cell-fate determinant in early-stage dividing colon cancer stem cells (CCSCs). In pair-cell assays, miR-34a distributes at high levels in differentiating progeny, whereas low levels of miR-34a demarcate self-renewing CCSCs. Moreover, miR-34a loss of function and gain of function alter the balance between self-renewal versus differentiation both in vitro and in vivo. Mechanistically, miR-34a sequesters Notch1 mRNA to generate a sharp threshold response where a bimodal Notch signal specifies the choice between self-renewal and differentiation. In contrast, the canonical cell-fate determinant Numb regulates Notch levels in a continuously graded manner. Altogether, our findings highlight a unique microRNA-regulated mechanism that converts noisy input into a toggle switch for robust cell-fate decisions in CCSCs.
Collapse
Affiliation(s)
- Pengcheng Bu
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853, USA
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Kai-Yuan Chen
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Joyce Huan Chen
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Lihua Wang
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853, USA
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jewell Walters
- Departments of Medicine, Genetic Medicine and Surgery, Weill Cornell Medical College, New York, NY 10021
| | - Yong Jun Shin
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Julian P. Goerger
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jian Sun
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10021, USA
| | - Mavee Witherspoon
- Departments of Medicine, Genetic Medicine and Surgery, Weill Cornell Medical College, New York, NY 10021
| | - Nikolai Rakhilin
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jiahe Li
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Herman Yang
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jeff Milsom
- Departments of Medicine, Genetic Medicine and Surgery, Weill Cornell Medical College, New York, NY 10021
| | - Sang Lee
- Departments of Medicine, Genetic Medicine and Surgery, Weill Cornell Medical College, New York, NY 10021
| | - Warren Zipfel
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Moonsoo M. Jin
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Zeynep H. Gümüşcedil
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10021, USA
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA
| | - Steven M. Lipkin
- Departments of Medicine, Genetic Medicine and Surgery, Weill Cornell Medical College, New York, NY 10021
| | - Xiling Shen
- School of Electrical and Computer Engineering, Cornell University, Ithaca, NY 14853, USA
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
153
|
Kamposioras K, Pentheroudakis G, Pavlidis N. Exploring the biology of cancer of unknown primary: breakthroughs and drawbacks. Eur J Clin Invest 2013; 43:491-500. [PMID: 23480555 DOI: 10.1111/eci.12062] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 02/05/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cancer of unknown primary (CUP) ranks among the ten most common malignancies worldwide. Cancer of unknown primary presents as disseminated disease, has a dismal prognosis and remains a diagnosis of exclusion. The natural history and biology of the disease is poorly understood, and efforts are focused on identifying the specific 'CUP signature'. MATERIALS AND METHODS We collected and analysed all published research in the biology of CUP from 1974 till present (Medline, Embase, ASCO and ESMO Congresses). RESULTS Current scientific evidence suggests that aneuploidy and karyotype changes are frequent, while more subtle molecular aberrations, such as epidermal growth factor receptor family proteins, cKit/PDGFR are frequently overexpressed, although without prognostic significance. Loss of function of tumour suppressor genes, active angiogenesis, a hypoxic genetic programme and a mesenchymal transitory phenotype have been reported in CUP and may be indicative of unfavourable prognosis. Molecular pathway analyses have identified various biomolecules impacting on survival (pAKT, pMAPK, c-Met, p21 and pPRS6). Finally, circulating tumour cells have recently been reported as a frequent phenomenon in CUP. CONCLUSIONS Overall, advances in understanding CUP biology have been weak and the application of gene expression profiling failed to identify an as yet elusive 'CUP molecular signature'. MicroRNA, epigenetic and proteomic studies are warranted to better characterize the biological profile of CUP and unravel its mystery.
Collapse
|
154
|
The cancer stem cell hypothesis applied to oral carcinoma. Oral Oncol 2013; 49:738-46. [PMID: 23642758 DOI: 10.1016/j.oraloncology.2013.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 04/09/2013] [Accepted: 04/09/2013] [Indexed: 12/14/2022]
Abstract
It has been proposed that the development of tumors is based exclusively on the activity of cancer stem cells (CSCs) leading to a new model of carcinogenesis, the CSC hypothesis, in opposition to the conventional model of clonal evolution. The new model may help to explain the high mortality of oral cancer, unchanged over the past decades, the low response to treatment and the tendency of oral squamous cell carcinoma (OSCC) patients to develop multiple tumors. However, a more profound understanding of the molecular pathways involved in maintaining the stem cell (SC) state and of their alterations is required to elucidate the mechanisms underlying the development of tumors and metastatic spread, but research into SC biopathology is hampered by the lack of specific markers for identifying SCs and CSCs in tissues and for establishing topographic relationships with their lineage. We review current knowledge on stem cells in relation to oral cancer, including their possible origins, focusing on the CSC hypothesis of oral tumorigenesis and attempts being made to identify oral stem cells.
Collapse
|
155
|
Galatro TFDA, Uno M, Oba-Shinjo SM, Almeida AN, Teixeira MJ, Rosemberg S, Marie SKN. Differential expression of ID4 and its association with TP53 mutation, SOX2, SOX4 and OCT-4 expression levels. PLoS One 2013; 8:e61605. [PMID: 23613880 PMCID: PMC3628974 DOI: 10.1371/journal.pone.0061605] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 03/11/2013] [Indexed: 02/07/2023] Open
Abstract
Inhibitor of DNA Binding 4 (ID4) is a member of the helix-loop-helix ID family of transcription factors, mostly present in the central nervous system during embryonic development, that has been associated with TP53 mutation and activation of SOX2. Along with other transcription factors, ID4 has been implicated in the tumorigenic process of astrocytomas, contributing to cell dedifferentiation, proliferation and chemoresistance. In this study, we aimed to characterize the ID4 expression pattern in human diffusely infiltrative astrocytomas of World Health Organization (WHO) grades II to IV of malignancy (AGII-AGIV); to correlate its expression level to that of SOX2, SOX4, OCT-4 and NANOG, along with TP53 mutational status; and to correlate the results with the clinical end-point of overall survival among glioblastoma patients. Quantitative real time PCR (qRT-PCR) was performed in 130 samples of astrocytomas for relative expression, showing up-regulation of all transcription factors in tumor cases. Positive correlation was found when comparing ID4 relative expression of infiltrative astrocytomas with SOX2 (r = 0.50; p<0.005), SOX4 (r = 0.43; p<0.005) and OCT-4 (r = 0.39; p<0.05). The results from TP53 coding exon analysis allowed comparisons between wild-type and mutated status only in AGII cases, demonstrating significantly higher levels of ID4, SOX2 and SOX4 in mutated cases (p<0.05). This pattern was maintained in secondary GBM and further confirmed by immunohistochemistry, suggesting a role for ID4, SOX2 and SOX4 in early astrocytoma tumorigenesis. Combined hyperexpression of ID4, SOX4 and OCT-4 conferred a much lower (6 months) median survival than did hypoexpression (18 months). Because both ID4 alone and a complex of SOX4 and OCT-4 activate SOX2 transcription, it is possible that multiple activation of SOX2 impair the prognosis of GBM patients. These observational results of associated expression of ID4 with SOX4 and OCT-4 may be used as a predictive factor of prognosis upon further confirmation in a larger GBM series.
Collapse
|
156
|
Singh SR. Gastric cancer stem cells: a novel therapeutic target. Cancer Lett 2013; 338:110-9. [PMID: 23583679 DOI: 10.1016/j.canlet.2013.03.035] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 03/25/2013] [Accepted: 03/30/2013] [Indexed: 12/14/2022]
Abstract
Gastric cancer remains one of the leading causes of global cancer mortality. Multipotent gastric stem cells have been identified in both mouse and human stomachs, and they play an essential role in the self-renewal and homeostasis of gastric mucosa. There are several environmental and genetic factors known to promote gastric cancer. In recent years, numerous in vitro and in vivo studies suggest that gastric cancer may originate from normal stem cells or bone marrow-derived mesenchymal cells, and that gastric tumors contain cancer stem cells. Cancer stem cells are believed to share a common microenvironment with normal niche, which play an important role in gastric cancer and tumor growth. This mini-review presents a brief overview of the recent developments in gastric cancer stem cell research. The knowledge gained by studying cancer stem cells in gastric mucosa will support the development of novel therapeutic strategies for gastric cancer.
Collapse
Affiliation(s)
- Shree Ram Singh
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
157
|
Response to stress in early tumor colonization modulates switching of CD133-positive and CD133-negative subpopulations in a human metastatic colon cancer cell line, SW620. PLoS One 2013; 8:e61133. [PMID: 23577199 PMCID: PMC3618272 DOI: 10.1371/journal.pone.0061133] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 03/05/2013] [Indexed: 02/07/2023] Open
Abstract
According to the cancer stem cell (CSC) model, higher CD133 expression in tumor tissue is associated with metastasis and poor prognosis in colon cancer. As such, the CD133-positive (CD133+) subpopulation of cancer cells is believed to play a central role in tumor development and metastatic progression. Although CD133+ cells are believed to display more CSC-like behavior and be solely responsible for tumor colonization, recent research indicates that CD133− cells from metastatic colon tumors not only also possess colonization capacity but also promote the growth of larger tumors in a mouse model than CD133+ cells, suggesting that an alternative mechanism of metastasis exists. This study investigated this possibility by examining the cell viability, tumorigenicity, and proliferation and growth capacity of the CD133+ and CD133− subpopulations of the SW620 cell line, a human metastatic colon cancer cell line, in both an in vitro cell model and an in vivo mouse model. While both SW620 CD133− and SW620CD133+ cells were found to engage in bidirectional cell-type switching in reaction to exposure to environmental stressors, including hypoxia, a cell adhesion-free environment, and extracellular matrix stimulation, both in vitro and in vivo, CD133− cells were found to have a growth advantage during early colonization due to their greater resistance to proliferation inhibition. Based on these findings, a hypothetical model in which colon cancer cells engage in cell-type switching in reaction to exposure to environmental stressors is proposed. Such switching may provide a survival advantage during early colonization, as well as that explain previous conflicting observations.
Collapse
|
158
|
Giudice FS, Pinto DS, Nör JE, Squarize CH, Castilho RM. Inhibition of histone deacetylase impacts cancer stem cells and induces epithelial-mesenchyme transition of head and neck cancer. PLoS One 2013; 8:e58672. [PMID: 23527004 PMCID: PMC3603970 DOI: 10.1371/journal.pone.0058672] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 02/05/2013] [Indexed: 12/30/2022] Open
Abstract
The genome is organized and packed into the nucleus through interactions with core histone proteins. Emerging evidence suggests that tumors are highly responsive to epigenetic alterations that induce chromatin-based events and dynamically influence tumor behavior. We examined chromatin organization in head and neck squamous cell carcinoma (HNSCC) using acetylation levels of histone 3 as a marker of chromatin compaction. Compared to control oral keratinocytes, we found that HNSCC cells are hypoacetylated and that microenvironmental cues (e.g., microvasculature endothelial cells) induce tumor acetylation. Furthermore, we found that chemical inhibition of histone deacetylases (HDAC) reduces the number of cancer stem cells (CSC) and inhibits clonogenic sphere formation. Paradoxically, inhibition of HDAC also induced epithelial-mesenchymal transition (EMT) in HNSCC cells, accumulation of BMI-1, an oncogene associated with tumor aggressiveness, and expression of the vimentin mesenchymal marker. Importantly, we observed co-expression of vimentin and acetylated histone 3 at the invasion front of human HNSCC tumor tissues. Collectively, these findings suggest that environmental cues, such as endothelial cell-secreted factors, modulate tumor plasticity by limiting the population of CSC and inducing EMT. Therefore, inhibition of HDAC may constitute a novel strategy to disrupt the population of CSC in head and neck tumors to create a homogeneous population of cancer cells with biologically defined signatures and predictable behavior.
Collapse
Affiliation(s)
- Fernanda S. Giudice
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, United States of America
- Department of Oral Pathology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Decio S. Pinto
- Department of Oral Pathology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Jacques E. Nör
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Cristiane H. Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, United States of America
| | - Rogerio M. Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
159
|
Affiliation(s)
- Martin E Fernandez-Zapico
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
160
|
Pathways to breast cancer recurrence. ISRN ONCOLOGY 2013; 2013:290568. [PMID: 23533807 PMCID: PMC3603357 DOI: 10.1155/2013/290568] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 01/17/2013] [Indexed: 12/22/2022]
Abstract
Breast cancer remains a deadly disease, even with all the recent technological advancements. Early intervention has made an impact, but an overwhelmingly large number of breast cancer patients still live under the fear of “recurrent” disease. Breast cancer recurrence is clinically a huge problem and one that is largely not well understood. Over the years, a number of factors have been studied with an overarching aim of being able to prognose recurrent disease. This paper attempts to provide an overview of our current knowledge of breast cancer recurrence and its associated challenges. Through a survey of the literature on cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT), various signaling pathways such as Notch/Wnt/hedgehog, and microRNAs (miRNAs), we also examine the hypotheses that are currently under investigation for the prevention of breast cancer recurrence.
Collapse
|
161
|
|
162
|
Scherzed A, Hackenberg S, Froelich K, Rak K, Technau A, Radeloff A, Nöth U, Koehler C, Hagen R, Kleinsasser N. Effects of salinomycin on human bone marrow-derived mesenchymal stem cells in vitro. Toxicol Lett 2013; 218:207-14. [PMID: 23410960 DOI: 10.1016/j.toxlet.2013.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 01/29/2013] [Accepted: 02/01/2013] [Indexed: 01/22/2023]
Abstract
Various hypotheses on the origin of cancer stem cells (CSCs) exist, including that CSCs develop from transformed human bone marrow mesenchymal stem cells (hBMSC). Since the polyether antibiotic salinomycin selectively kills CSCs, the present study aims to elucidate the effects of salinomycin on normal hBMSC. The immunophenotype of hBMSC after salinomycin exposure was observed by flow cytometry. The multi-differentiation capacity of hBMSC was evaluated by Oil Red O and van Kossa staining. Cytotoxic effects of salinomycin were monitored by the [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] (MTT) assay. Furthermore, spheroid formation and migration capacity were assessed. There were no differences in the immunophenotype and multi-differentiation capacity of hBMSC induced by salinomycin treatment. Cytotoxic effects were observed at concentrations of 30 μM and above. Neither the migration capability nor the ability to form spheroids was affected. Essential functional properties of hBMSC were unaffected by salinomycin. However, dose-dependent cytotoxicity effects could be observed. Overall, low dose salinomycin showed no negative effects on hBMSC. Since mesenchymal stem cells from various sources respond differently, further in vitro studies are needed to clarify the effect of salinomycin on tissue-specific stem cells.
Collapse
Affiliation(s)
- A Scherzed
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University of Wuerzburg, Josef-Schneider Str. 11, 97080 Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|